Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.152
Filtrar
1.
Sci Rep ; 12(1): 2306, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35145145

RESUMEN

Myocardin-related transcription factor A (MRTF-A), a coactivator of serum response factor (SRF), regulates the expression of many cytoskeletal genes in response to cytoplasmic and nuclear actin dynamics. Here we describe a novel mechanism to regulate MRTF-A activity within the nucleus by showing that lamina-associated polypeptide 2α (Lap2α), the nucleoplasmic isoform of Lap2, is a direct binding partner of MRTF-A, and required for the efficient expression of MRTF-A/SRF target genes. Mechanistically, Lap2α is not required for MRTF-A nuclear localization, unlike most other MRTF-A regulators, but is required for efficient recruitment of MRTF-A to its target genes. This regulatory step takes place prior to MRTF-A chromatin binding, because Lap2α neither interacts with, nor specifically influences active histone marks on MRTF-A/SRF target genes. Phenotypically, Lap2α is required for serum-induced cell migration, and deregulated MRTF-A activity may also contribute to muscle and proliferation phenotypes associated with loss of Lap2α. Our studies therefore add another regulatory layer to the control of MRTF-A-SRF-mediated gene expression, and broaden the role of Lap2α in transcriptional regulation.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/fisiología , Transactivadores/genética , Transactivadores/metabolismo , Actinas/metabolismo , Animales , Movimiento Celular/genética , Cromatina , Citoplasma/metabolismo , Citoesqueleto/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Células 3T3 NIH , Unión Proteica/genética , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Transactivadores/fisiología , Transcripción Genética/genética
2.
Sci Rep ; 12(1): 1251, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35075262

RESUMEN

Staphylococcus aureus is an opportunistic, pathogenic bacteria that causes significant morbidity and mortality. As antibiotic resistance by S. aureus continues to be a serious concern, developing novel drug therapies to combat these infections is vital. Quorum sensing inhibitors (QSI) dampen S. aureus virulence and facilitate clearance by the host immune system by blocking quorum sensing signaling that promotes upregulation of virulence genes controlled by the accessory gene regulator (agr) operon. While QSIs have shown therapeutic promise in mouse models of S. aureus skin infection, their further development has been hampered by the suggestion that agr inhibition promotes biofilm formation. In these studies, we investigated the relationship between agr function and biofilm growth across various S. aureus strains and experimental conditions, including in a mouse model of implant-associated infection. We found that agr deletion was associated with the presence of increased biofilm only under narrow in vitro conditions and, crucially, was not associated with enhanced biofilm development or enhanced morbidity in vivo.


Asunto(s)
Proteínas Bacterianas/fisiología , Biopelículas/crecimiento & desarrollo , Staphylococcus aureus/fisiología , Transactivadores/fisiología , Animales , Técnicas de Cultivo , Femenino , Ratones Endogámicos BALB C , Percepción de Quorum
3.
Mol Oncol ; 16(2): 422-446, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34197030

RESUMEN

WW-domain-binding protein 2 (WBP2) is an oncogene that drives breast carcinogenesis through regulating Wnt, estrogen receptor (ER), and Hippo signaling. Recent studies have identified neoteric modes of action of WBP2 other than its widely recognized function as a transcriptional coactivator. Here, we identified a previously unexplored role of WBP2 in inflammatory signaling in breast cancer via an integrated proteogenomic analysis of The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA BRCA) dataset. WBP2 was shown to enhance the migration and invasion in triple-negative breast cancer (TNBC) cells especially under tumor necrosis factor alpha (TNF-α) stimulation. Molecularly, WBP2 potentiates TNF-α-induced nuclear factor kappa B (NF-κB) transcriptional activity and nuclear localization through aggrandizing ubiquitin-mediated proteasomal degradation of its upstream inhibitor, NF-κB inhibitor alpha (NFKBIA; also known as IκBα). We further demonstrate that WBP2 induces mRNA stability of beta-transducin repeat-containing E3 ubiquitin protein ligase (BTRC), which targets IκBα for ubiquitination and degradation. Disruption of IκBα rescued the impaired migratory and invasive phenotypes in WBP2-silenced cells, while loss of BTRC ameliorated WBP2-driven migration and invasion. Clinically, the WBP2-BTRC-IκBα signaling axis correlates with poorer prognosis in breast cancer patients. Our findings reveal a pivotal mechanism of WBP2 in modulating BTRC-IκBα-NF-κB pathway to promote TNBC aggressiveness.


Asunto(s)
FN-kappa B/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , ARN Mensajero/genética , Transactivadores/fisiología , Neoplasias de la Mama Triple Negativas/patología , Proteínas con Repetición de beta-Transducina/genética , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Femenino , Humanos , Inflamación/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo
4.
Mol Med Rep ; 25(1)2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34779499

RESUMEN

Our previous study revealed that metastasis­associated protein 1 (MTA1), which is expressed in vascular endothelial cells, acts as a tube formation promoting factor. The present study aimed to clarify the importance of MTA1 expression in tube formation using MTA1­knockout (KO) endothelial cells (MTA1­KO MSS31 cells). Tube formation was significantly suppressed in MTA1­KO MSS31 cells, whereas MTA1­overexpression MTA1­KO MSS31 cells regained the ability to form tube­like structures. In addition, western blotting analysis revealed that MTA1­KO MSS31 cells showed significantly higher levels of phosphorylation of non­muscle myosin heavy chain IIa, which resulted in suppression of tube formation. This effect was attributed to a decrease of MTA1/S100 calcium­binding protein A4 complex formation. Moreover, inhibition of tube formation in MTA1­KO MSS31 cells could not be rescued by stimulation with vascular endothelial growth factor (VEGF). These results demonstrated that MTA1 may serve as an essential molecule for angiogenesis in endothelial cells and be involved in different steps of the angiogenic process compared with the VEGF/VEGF receptor 2 pathway. The findings showed that endothelial MTA1 and its pathway may serve as promising targets for inhibiting tumor angiogenesis, further supporting the development of MTA1­based antiangiogenic therapies.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Células Cultivadas , China , Ratones , Morfogénesis/efectos de los fármacos , Cadenas Pesadas de Miosina/metabolismo , Metástasis de la Neoplasia/genética , Fosforilación , Proteínas Represoras/fisiología , Proteína de Unión al Calcio S100A4/metabolismo , Transactivadores/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Mediators Inflamm ; 2021: 6359652, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34924813

RESUMEN

Ellagic acid (EA) was reported to play protective roles in rheumatoid arthritis (RA). It was found that the level of metastasis-associated gene 1 (MTA1)/histone deacetylase 1 (HDAC1) protein complex was downregulated by polyphenols in several human disorders. Notably, inhibition of MTA1 or HDAC1 has anti-inflammatory effects on RA. Therefore, our study is aimed at investigating whether EA prevents RA progression through regulating the MTA1/HDAC1 complex. Herein, the human fibroblast-like synoviocyte (FLS) cell line MH7A was treated with TNF-α to induce an inflammation model in vitro and then incubated with different concentrations of EA. Western blot analysis showed that EA reduced MTA1 expression in a dose-dependent manner in MH7A cells. Then, TNF-α-treated MH7A cells were incubated with EA alone or together with MTA1 overexpression plasmid (pcDNA-MTA1), and we found that EA inhibited proliferation, inflammation cytokine levels, and oxidative stress marker protein levels and promoted apoptosis in MH7A cells, while MTA1 overexpression abolished these effects. Moreover, coimmunoprecipitation assay verified the interaction between MTA1 and HDAC1. EA downregulated the MTA1/HDAC1 complex in MH7A cells. MTA1 knockdown inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells, while HDAC1 overexpression reversed these effects. Moreover, chromatin immunoprecipitation assay indicated that EA inhibited HDAC1-mediated Nur77 deacetylation. Rescue experiments demonstrated that Nur77 knockdown reversed the effects of EA on MH7A cell biological behaviors. Additionally, EA treatment attenuated arthritis index, paw swelling, synovial hyperplasia, and inflammation in collagen-induced arthritis (CIA) rats. In conclusion, EA inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells and alleviated the severity of RA in CIA rats though downregulating MTA1/HDAC1 complex and promoting HDAC1 deacetylation-mediated Nur77 expression.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Ácido Elágico/farmacología , Histona Desacetilasa 1/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Acetilación , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Histona Desacetilasa 1/fisiología , Humanos , Masculino , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Proteínas Represoras/fisiología , Transactivadores/fisiología
6.
J BUON ; 26(4): 1422-1431, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34565000

RESUMEN

PURPOSE: Radiotherapy is the most commonly selective medical treatment for non-small cell lung cancer (NSCLC) and the multiple underlying mechanisms are considered as the effectively theoretical foundation. Herein, we investigated the effects of let-7a targets Rsf-1 on modulating the radiotherapy response in NSCLC cells by Ras-MAPK pathway. METHODS: A549 cells were divided into different groups to investigate the role of let-7a and Rsf-1 on the radiotherapy response. The expression of let-7a and Rsf-1 were detected by RT-PCR. Bioinformatic analysis indicated that Rsf-1 is the target of let-7a. The binding site of let-7a in the Rsf-1 3'UTR was detected based on double luciferase reporter assay and Western blot. The cell variability and proliferation were assessed by MTT and colony formation assay. The expression levels of Ras-MARK signaling pathway related proteins were assessed by RT-PCR. RESULTS: RT-PCR results showed that radiotherapy could up-regulate the expression of let-7a, thereby reducing the expression of Rsf-1, and the correlation between the two factors was negatively correlated. At the same time, let-7a overexpression and Rsf-1 silencing could further reduce the activity of A549 cells after radiotherapy, have an inhibitory effect on cell proliferation and inhibit the expression of related proteins in the Ras-MAPK pathway. CONCLUSIONS: Rsf-1 is the target of Let-7a. The present study provides evidence that let-7a targeting Rsf-1 can modulate radiotherapy response in NSCLC cells through Ras-MAPK pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Neoplasias Pulmonares/radioterapia , MicroARNs/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteínas Nucleares/fisiología , Transducción de Señal/fisiología , Transactivadores/fisiología , Proteínas ras/fisiología , Humanos , Células Tumorales Cultivadas
7.
Curr Microbiol ; 78(11): 3936-3944, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34522977

RESUMEN

Our study attempted to explore the mechanism underlying the role of LuxR family transcriptional regulator abaR in biofilm formation by Acinetobacter baumannii. The abaR gene was knocked out in ATCC 17978 strain using homologous recombination method. The growth curve and biofilm formation were measured in the wild type and abaR gene knockdown strains. Transcriptome sequencing was performed in the wild type and abaR gene knockdown strains following 8 h of culture. The growth curve in the abaR gene knockdown strain was similar to that of the wild-type strain. Biofilm formation significantly declined in the abaR gene knockdown strain at 8 and 48 h after culture. A total of 137 differentially expressed genes (DEGs) were obtained including 20 downregulated DEGs and 117 upregulated DEGs. Genes with differential expression were closely related to viral procapsid maturation (GO:0046797), acetoin catabolism (GO:0045150), carbon metabolism (ko01200), and the glycolysis/gluconeogenesis (ko00010)-related pathways. The results of the eight verified expression DEGs were consistent with the results predicted by bioinformatics. AbaR gene knockdown significantly affected biofilm formation by A. baumannii ATCC 17978 strain. The glycolysis/gluconeogenesis pathways were significantly dysregulated and induced by abaR gene knockdown in A. baumannii.


Asunto(s)
Acinetobacter baumannii , Proteínas Represoras/fisiología , Transactivadores/fisiología , Acinetobacter baumannii/genética , Biopelículas , Proteínas Represoras/genética , Transactivadores/genética
8.
Mol Neurodegener ; 16(1): 64, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526055

RESUMEN

BACKGROUND: Human genetic association studies point to immune response and lipid metabolism, in addition to amyloid-beta (Aß) and tau, as major pathways in Alzheimer's disease (AD) etiology. Accumulating evidence suggests that chronic neuroinflammation, mainly mediated by microglia and astrocytes, plays a causative role in neurodegeneration in AD. Our group and others have reported early and dramatic losses of brain sulfatide in AD cases and animal models that are mediated by ApoE in an isoform-dependent manner and accelerated by Aß accumulation. To date, it remains unclear if changes in specific brain lipids are sufficient to drive AD-related pathology. METHODS: To study the consequences of CNS sulfatide deficiency and gain insights into the underlying mechanisms, we developed a novel mouse model of adult-onset myelin sulfatide deficiency, i.e., tamoxifen-inducible myelinating glia-specific cerebroside sulfotransferase (CST) conditional knockout mice (CSTfl/fl/Plp1-CreERT), took advantage of constitutive CST knockout mice (CST-/-), and generated CST/ApoE double knockout mice (CST-/-/ApoE-/-), and assessed these mice using a broad range of methodologies including lipidomics, RNA profiling, behavioral testing, PLX3397-mediated microglia depletion, mass spectrometry (MS) imaging, immunofluorescence, electron microscopy, and Western blot. RESULTS: We found that mild central nervous system (CNS) sulfatide losses within myelinating cells are sufficient to activate disease-associated microglia and astrocytes, and to increase the expression of AD risk genes (e.g., Apoe, Trem2, Cd33, and Mmp12), as well as previously established causal regulators of the immune/microglia network in late-onset AD (e.g., Tyrobp, Dock, and Fcerg1), leading to chronic AD-like neuroinflammation and mild cognitive impairment. Notably, neuroinflammation and mild cognitive impairment showed gender differences, being more pronounced in females than males. Subsequent mechanistic studies demonstrated that although CNS sulfatide losses led to ApoE upregulation, genetically-induced myelin sulfatide deficiency led to neuroinflammation independently of ApoE. These results, together with our previous studies (sulfatide deficiency in the context of AD is mediated by ApoE and accelerated by Aß accumulation) placed both Aß and ApoE upstream of sulfatide deficiency-induced neuroinflammation, and suggested a positive feedback loop where sulfatide losses may be amplified by increased ApoE expression. We also demonstrated that CNS sulfatide deficiency-induced astrogliosis and ApoE upregulation are not secondary to microgliosis, and that astrogliosis and microgliosis seem to be driven by activation of STAT3 and PU.1/Spi1 transcription factors, respectively. CONCLUSION: Our results strongly suggest that sulfatide deficiency is an important contributor and driver of neuroinflammation and mild cognitive impairment in AD pathology.


Asunto(s)
Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Trastornos de la Memoria/metabolismo , Vaina de Mielina/química , Enfermedades Neuroinflamatorias/metabolismo , Sulfoglicoesfingolípidos/metabolismo , Edad de Inicio , Enfermedad de Alzheimer/etiología , Aminopiridinas/toxicidad , Animales , Apolipoproteínas E/metabolismo , Química Encefálica , Sistema Nervioso Central/metabolismo , Disfunción Cognitiva/etiología , Perfilación de la Expresión Génica , Gliosis/metabolismo , Humanos , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Noqueados para ApoE , Prueba del Laberinto Acuático de Morris , Neuroglía/enzimología , Neuroglía/fisiología , Enfermedades Neuroinflamatorias/etiología , Prueba de Campo Abierto , Proteínas Proto-Oncogénicas/fisiología , Pirroles/toxicidad , Factor de Transcripción STAT3/fisiología , Sulfoglicoesfingolípidos/análisis , Sulfotransferasas/deficiencia , Transactivadores/fisiología
9.
Cancer Lett ; 523: 57-71, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34563641

RESUMEN

High fluence low-level laser (HF-LLL), a mitochondria-targeted tumour phototherapy, results in oxidative damage and apoptosis of tumour cells, as well as damage to normal tissue. To circumvent this, the therapeutic effect of low fluence LLL (LFL), a non-invasive and drug-free therapeutic strategy, was identified for tumours and the underlying molecular mechanisms were investigated. We observed that LFL enhanced antigen-specific immune response of macrophages and dendritic cells by upregulating MHC class II, which was induced by mitochondrial reactive oxygen species (ROS)-activated signalling, suppressing tumour growth in both CD11c-DTR and C57BL/6 mice. Mechanistically, LFL upregulated MHC class II in an MHC class II transactivator (CIITA)-dependent manner. LFL-activated protein kinase C (PKC) promoted the nuclear translocation of CIITA, as inhibition of PKC attenuated the DNA-binding efficiency of CIITA to MHC class II promoter. CIITA mRNA and protein expression also improved after LFL treatment, characterised by direct binding of Src and STAT1, and subsequent activation of STAT1. Notably, scavenging of ROS downregulated LFL-induced Src and PKC activation and antagonised the effects of LFL treatment. Thus, LFL treatment altered the adaptive immune response via the mitochondrial ROS-activated signalling pathway to control the progress of neoplastic disease.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/inmunología , Terapia por Luz de Baja Intensidad/métodos , Neoplasias Experimentales/terapia , Proteína Quinasa C/fisiología , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/inmunología , Familia-src Quinasas/fisiología , Transporte Activo de Núcleo Celular , Animales , Presentación de Antígeno , Células Dendríticas/fisiología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Proteínas Nucleares/fisiología , Factor de Transcripción STAT1/fisiología , Transactivadores/fisiología
10.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34507985

RESUMEN

Episodic events are frequently consolidated into labile memory but are not necessarily transferred to persistent long-term memory (LTM). Regulatory mechanisms leading to LTM formation are poorly understood, however, especially at the resolution of identified neurons. Here, we demonstrate enhanced LTM following aversive olfactory conditioning in Drosophila when the transcription factor cyclic AMP response element binding protein A (CREBA) is induced in just two dorsal-anterior-lateral (DAL) neurons. Our experiments show that this process is regulated by protein-gene interactions in DAL neurons: (1) crebA transcription is induced by training and repressed by crebB overexpression, (2) CREBA bidirectionally modulates LTM formation, (3) crebA overexpression enhances training-induced gene transcription, and (4) increasing membrane excitability enhances LTM formation and gene expression. These findings suggest that activity-dependent gene expression in DAL neurons during LTM formation is regulated by CREB proteins.


Asunto(s)
Proteína de Unión al Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , Memoria a Largo Plazo/fisiología , Transactivadores/metabolismo , Animales , Condicionamiento Clásico/fisiología , Condicionamiento Psicológico , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Proteína de Unión al Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión al Elemento de Respuesta al AMP Cíclico/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster , Femenino , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Masculino , Neuronas/metabolismo , Neuronas/fisiología , Percepción Olfatoria/fisiología , Olfato/fisiología , Transactivadores/fisiología
11.
Genes (Basel) ; 12(7)2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34356097

RESUMEN

Acquisition of cellular fate during development is initiated and maintained by well-coordinated patterns of gene expression that are dictated by the epigenetic landscape and genome organization in the nucleus. While the epigenetic marks that mediate developmental gene expression patterns during organogenesis have been well studied, less is known about how epigenetic marks influence nuclear organization during development. This study examines the relationship between nuclear structure, chromatin accessibility, DNA methylation, and gene expression during hepatic outgrowth in zebrafish larvae. We investigate the relationship between these features using mutants that lack DNA methylation. Hepatocyte nuclear morphology was established coincident with hepatocyte differentiation at 80 h post-fertilization (hpf), and nuclear shape and size continued to change until the conclusion of outgrowth and morphogenesis at 120 hpf. Integrating ATAC-Seq analysis with DNA methylation profiling of zebrafish livers at 120 hpf showed that closed and highly methylated chromatin occupies most transposable elements and that open chromatin correlated with gene expression. DNA hypomethylation, due to mutation of genes encoding ubiquitin-like, containing PHD and RING Finger Domains 1 (uhrf1) and DNA methyltransferase (dnmt1), did not block hepatocyte differentiation, but had dramatic effects on nuclear organization. Hepatocytes in uhrf1 mutants have large, deformed nuclei with multiple nucleoli, downregulation of nucleolar genes, and a complete lack of the nuclear lamina. Loss of lamin B2 staining was phenocopied by dnmt1 mutation. Together, these data show that hepatocyte nuclear morphogenesis coincides with organ morphogenesis and outgrowth, and that DNA methylation directs chromatin organization, and, in turn, hepatocyte nuclear shape and size during liver development.


Asunto(s)
Hepatocitos/metabolismo , Transactivadores/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Diferenciación Celular/genética , Núcleo Celular/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN/genética , Epigénesis Genética/genética , Expresión Génica/genética , Larva/genética , Hígado/embriología , Hígado/metabolismo , Organogénesis/genética , Transactivadores/genética , Transactivadores/fisiología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
12.
Theranostics ; 11(17): 8362-8378, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34373747

RESUMEN

Rationale: Hepatitis B x protein (HBx) is required to initiate and maintain the replication of hepatitis B virus (HBV). Protein arginine methyltransferases 5 (PRMT5) negatively regulates HBV transcription. WD repeat domain 77 protein (WDR77) greatly enhances the methyltransferase activity of PRMT5. However, the role of WDR77 in the modulation of cccDNA transcription and HBV replication is poorly understood. In this study, we investigated the mechanism by which HBx modulated HBV replication involving WDR77 in the liver. Methods: A human liver-chimeric mouse model was established. Immunohistochemistry (IHC) staining, Western blot analysis, Southern blot analysis, Northern blot analysis, immunofluorescence assays, ELISA, RT-qPCR, CoIP assays, and ChIP assays were performed in human liver-chimeric mouse model, primary human hepatocytes (PHHs), HepG2-NTCP, dHepaRG and HepG2 cell lines. Results: HBV infection and HBx expression remarkably reduced the protein levels of WDR77 in human liver-chimeric mice and HepG2-NTCP cells. WDR77 restricted cccDNA transcription and HBV replication in PHHs and HepG2-NTCP cells. Mechanically, WDR77 enhanced PRMT5-triggered symmetric dimethylation of arginine 3 on H4 (H4R3me2s) on the cccDNA minichromosome to control cccDNA transcription. HBx drove the cellular DDB1-containing E3 ubiquitin ligase to degrade WDR77 through recruiting WDR77, leading to the disability of methyltransferase activity of PRMT5. Thus, HBx promoted HBV replication by driving a positive feedback loop of HBx-DDB1/WDR77/PRMT5/H4R3me2s/cccDNA/HBV/HBx in the liver. Conclusions: HBx attenuates the WDR77-mediated HBV repression by driving DDB1-induced WDR77 degradation in the liver. Our finding provides new insights into the mechanism by which HBx enhances HBV replication in the liver.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Adulto , Anciano , Animales , Quimera , China , ADN Viral/genética , Proteínas de Unión al ADN/fisiología , Femenino , Células Hep G2 , Hepatitis B/genética , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/patogenicidad , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Proteína-Arginina N-Metiltransferasas/metabolismo , Transactivadores/fisiología , Factores de Transcripción/fisiología , Transcripción Genética/genética , Proteínas Reguladoras y Accesorias Virales/fisiología , Replicación Viral
13.
J Cell Mol Med ; 25(18): 8645-8661, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34347392

RESUMEN

Myocardin-related transcription factor-A/serum response factor (MRTF-A/SRF), a well-known transcriptional programme, has been proposed to play crucial roles in skeletal muscle development and function. However, whether MRTF-A participates in muscle regeneration and the molecular mechanisms are not completely understood. Here, we show that MRTF-A levels are highly correlated with myogenic genes using a RNA-seq assay, which reveal that MRTF-A knockdown in C2C12 cells significantly reduces PAX7 expression. Subsequent in vitro and in vivo data show that MRTF-A and PAX7 present identical expression patterns during myoblast differentiation and CTX-induced muscle injury and repair. Remarkably, MRTF-A overexpression promotes myoblast proliferation, while inhibiting cell differentiation and the expression of MyoD and MyoG. MRTF-A loss of function produces the opposite effect. Moreover, mice with lentivirus (MRTF-A) injection possesses more PAX7+ satellite cells, but less differentiating MyoD+ and MyoG+ cells, leading subsequently to diminished muscle regeneration. Our mechanistic results reveal that MRTF-A contributes to PAX7-mediated myoblast self-renewal, proliferation, and differentiation by binding to its distal CArG box. Overall, we propose that MRTF-A functions as a novel PAX7 regulator upon myoblast commitment to differentiation, which could provide pathways for dictating muscle stem cell fate and open new avenues to explore stem cell-based therapy for muscle degenerative diseases.


Asunto(s)
Desarrollo de Músculos , Músculos/metabolismo , Mioblastos , Transactivadores/fisiología , Animales , Línea Celular , Ratones , Ratones Endogámicos C57BL , Mioblastos/citología , Mioblastos/metabolismo , Regeneración
14.
Life Sci Alliance ; 4(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34117091

RESUMEN

WBP2 is an emerging oncoprotein with diverse functions in breast tumorigenesis via regulating Wnt, epidermal growth factor receptor, estrogen receptor, and Hippo. Recently, evidence shows that WBP2 is tightly regulated by the components of the miRNA biogenesis machinery such as DGCR8 and Dicer via producing both WBP2's 3'UTR and coding DNA sequence-targeting miRNAs. This led us to hypothesize that WBP2 could provide a feedback loop to the biogenesis of its key upstream regulators by regulating the microprocessor complex activity. Indeed, WBP2 suppressed microprocessor activity by blocking the processing of pri-miRNAs to pre-miRNAs. WBP2 negatively regulated the assembly of the microprocessor complex via physical interactions with its components. Meta-analyses suggest that microprocessor complex components, in particular DGCR8, DDX5, and DEAD-Box Helicase17 (DDX17), have tumor-suppressive properties. 2D and 3D in vitro proliferation assays revealed that WBP2 blocked the tumor-suppressive properties of DGCR8, a key component of the microprocessor complex. In conclusion, WBP2 is a novel regulator of miRNA biogenesis that is a known dysregulated pathway in breast tumorigenesis. The reregulation of miRNA biogenesis machinery via targeting WBP2 protein may have implications in breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/genética , MicroARNs/biosíntesis , Transactivadores/metabolismo , Neoplasias de la Mama/metabolismo , Carcinogénesis/genética , Línea Celular Tumoral , ARN Helicasas DEAD-box/metabolismo , Femenino , Humanos , MicroARNs/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III/metabolismo , Transactivadores/fisiología
15.
Cell Death Dis ; 12(6): 525, 2021 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-34023861

RESUMEN

While pregnancy is known to reduce a woman's life-long risk of breast cancer, clinical data suggest that it can specifically promote HER2 (human EGF receptor 2)-positive breast cancer subtype (HER2+ BC). HER2+ BC, characterized by amplification of HER2, comprises about 20% of all sporadic breast cancers and is more aggressive than hormone receptor-positive breast cancer (the majority of cases). Consistently with human data, pregnancy strongly promotes HER2+ BC in genetic mouse models. One proposed mechanism of this is post-pregnancy accumulation of PIMECs (pregnancy-identified mammary epithelial cells), tumor-initiating cells for HER2+ BC in mice. We previously showed that p63, a homologue of the tumor suppressor p53, is required to maintain the post-pregnancy number of PIMECs and thereby promotes HER2+ BC. Here we set to test whether p63 also affects the intrinsic tumorigenic properties of PIMECs. To this end, we FACS-sorted YFP-labeled PIMECs from p63+/-;ErbB2 and control p63+/+;ErbB2 females and injected their equal amounts into immunodeficient recipients. To our surprise, p63+/- PIMECs showed increased, rather than decreased, tumorigenic capacity in vivo, i.e., significantly accelerated tumor onset and tumor growth, as well as increased self-renewal in mammosphere assays and proliferation in vitro and in vivo. The underlying mechanism of these phenotypes seems to be a specific reduction of the tumor suppressor TAp63 isoform in p63+/- luminal cells, including PIMECs, with concomitant aberrant upregulation of the oncogenic ΔNp63 isoform, as determined by qRT-PCR and scRNA-seq analyses. In addition, scRNA-seq revealed upregulation of several cancer-associated (Il-4/Il-13, Hsf1/HSP), oncogenic (TGFß, NGF, FGF, MAPK) and self-renewal (Wnt, Notch) pathways in p63+/-;ErbB2 luminal cells and PIMECs per se. Altogether, these data reveal a complex role of p63 in PIMECs and pregnancy-associated HER2+ BC: maintaining the amount of PIMECs while suppressing their intrinsic tumorigenic capacity.


Asunto(s)
Neoplasias de la Mama/patología , Células Madre Neoplásicas/fisiología , Embarazo/fisiología , Transactivadores/fisiología , Animales , Mama/patología , Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Epiteliales/patología , Células Epiteliales/fisiología , Femenino , Genes erbB-2 , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Células Madre Neoplásicas/patología , Transactivadores/genética
16.
Theranostics ; 11(13): 6592-6606, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995678

RESUMEN

Purpose: Clinical success of cancer therapy is severely limited by drug resistance, attributed in large part to the loss of function of tumor suppressor genes (TSGs). Developing effective strategies to treat those tumors is challenging, but urgently needed in clinic. Experimental Design: MYOCD is a clinically relevant TSG in lung cancer patients. Our in vitro and in vivo data confirm its tumor suppressive function. Further analysis reveals that MYOCD potently inhibits stemness of lung cancer stem cells. Mechanistically, MYOCD localizes to TGFBR2 promoter region and thereby recruits PRMT5/MEP50 complex to epigenetically silence its transcription. Conclusions: NSCLC cells deficient of MYOCD are particularly sensitive to TGFBR kinase inhibitor (TGFBRi). TGFBRi and stemness inhibitor synergize with existing drugs to treat MYOCD deficient lung cancers. Our current work shows that loss of function of MYOCD creates Achilles' heels in lung cancer cells, which might be exploited in clinic.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas Nucleares/deficiencia , Receptor Tipo II de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Transactivadores/deficiencia , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación hacia Abajo , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Código de Histonas , Humanos , Neoplasias Pulmonares/genética , Metilación , Ratones Transgénicos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/patología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Transducción de Señal , Transactivadores/biosíntesis , Transactivadores/genética , Transactivadores/fisiología , Carga Tumoral
17.
Biochem Biophys Res Commun ; 558: 51-56, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33895551

RESUMEN

The quorum-sensing (QS) system between the phages and their hosts is important for the phage lysis-lysogeny decision. In Vibrio cholerae, the QS system consists of a LuxR-type receptor VqmA (VqmAVc) and an autoinducer molecule 3,5-dimethylpyrazin-2-ol (DPO). A VqmA homolog encoded by vibriophage VP882 (VqmAPhage) can intervene the host QS system via binding to both the host-produced DPO and its cognate promoter (Pqtip) to induce the phage lysogeny-to-lysis transition, whereas VqmAVc cannot influence the VqmAPhage-induced pathway, suggesting an asymmetry regulation. In this study, we report the crystal structure of VqmAPhage-DPO complex at 2.65 Å and reveal that the mechanism of DPO recognition is conserved in VqmA homologs. Besides, we identify a non-classical palindrome sequence in Pqtip, which can be effectively recognized by VqmAPhage but not VqmAVc. The sequence contains an interval longer than that in the vqmR promoter recognized by VqmAVc. In addition, the two DBD regions in the VqmAPhage dimer exhibit more relaxed architecture than that of the reported VqmAVc, which is likely to be in the conformation that may easily bind to target promoter containing a longer interval. In summary, our findings provide a structural and biochemical basis for the DBD-dependent DNA recognition in different promoter regions in the phage lysogeny-to-lysis decision communication system, and provide clues for developing phage therapies against Vibrio cholerae infection.


Asunto(s)
Bacteriófagos/genética , Percepción de Quorum/genética , Vibrio cholerae/virología , Bacteriólisis/genética , Bacteriólisis/fisiología , Bacteriófagos/patogenicidad , Bacteriófagos/fisiología , Cristalografía por Rayos X , Regulación Viral de la Expresión Génica , Genes Virales , Humanos , Lisogenia/genética , Lisogenia/fisiología , Modelos Moleculares , Regiones Promotoras Genéticas , Conformación Proteica , Percepción de Quorum/fisiología , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Transactivadores/química , Transactivadores/genética , Transactivadores/fisiología , Vibrio cholerae/fisiología
18.
Hum Cell ; 34(3): 918-931, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33813726

RESUMEN

Gallbladder carcinoma (GBC) is a lethal biliary tract malignant neoplasm. Patient-derived primary cancer cell lines (PDPCs) are appropriate models to explore biological characteristics and potential therapeutics; however, there is a lack of PDPCs in GBC. In this study, we aimed to establish and characterize the GBC PDPCs, and further investigated the intra-tumor heterogeneity (ITH). Multi-region sampling (3-9 regions) of the operable tumor tissue samples was used to establish PDPCs. Short tandem repeat genotyping for cell authentication and karyotyping was performed, followed by whole-exome sequencing and RNA sequencing to assess the ITH at the genetic and transcriptional levels, respectively. Thirty-eight PDPCs were successfully established from seven GBC patients and characterized. ITH was observed with a median of 38.3% mutations being heterogeneous (range, 26.6-59.4%) across all patients. Similar with other tumor types, TP53 mutations were always truncal. In addition, there were three genes, KMT2C, CDKN2A, and ARID1A, with truncal mutations in at least two patients. A median of 370 differentially expressed genes (DEGs) was identified per patient. Distinct expression patterns were observed between major histocompatibility complex (MHC) class I and II genes. We found the expression of MHC class II genes in the PDPC samples was closely regulated by CIITA, while that of MHC class I genes were not correlated with CIITA expression. The PDPCs established from GBC patients can serve as novel in vitro models to identify the ITH, which may pave a crucial molecular foundation for enhanced understanding of tumorigenesis and progression.


Asunto(s)
Carcinoma/genética , Carcinoma/patología , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/patología , Heterogeneidad Genética , Carcinogénesis/genética , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Genes MHC Clase I , Genes MHC Clase II , Humanos , Mutación , Proteínas Nucleares/fisiología , Transactivadores/fisiología , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética
19.
J Biol Chem ; 296: 100543, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33722605

RESUMEN

Myocardin-related transcription factor A (MRTFA) is a coactivator of serum response factor, a transcription factor that participates in several critical cellular functions including cell growth and apoptosis. MRTFA couples transcriptional regulation to actin cytoskeleton dynamics, and the transcriptional targets of the MRTFA-serum response factor complex include genes encoding cytoskeletal proteins as well as immediate early genes. Previous work has shown that MRTFA promotes the differentiation of many cell types, including various types of muscle cells and hematopoietic cells, and MRTFA's interactions with other protein partners broaden its cellular roles. However, despite being first identified as part of the recurrent t(1;22) chromosomal translocation in acute megakaryoblastic leukemia, the mechanisms by which MRTFA functions in malignant hematopoiesis have yet to be defined. In this review, we provide an in-depth examination of the structure, regulation, and known functions of MRTFA with a focus on hematopoiesis. We conclude by identifying areas of study that merit further investigation.


Asunto(s)
Hematopoyesis/fisiología , Transactivadores/fisiología , Animales , Humanos , Unión Proteica , Procesamiento Proteico-Postraduccional , ARN Mensajero/genética , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Transactivadores/química , Transactivadores/genética , Transcripción Genética
20.
Mol Plant Microbe Interact ; 34(8): 952-961, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33779205

RESUMEN

LuxR-type transcriptional regulators are essential for many physiological processes in bacteria, including pathogenesis. Acidovorax citrulli is a seedborne bacterial pathogen responsible for bacterial fruit blotch, which causes great losses in melon and watermelon worldwide. However, the LuxR-type transcriptional factors in A. citrulli have not been well studied, except for the previously reported LuxR-type regulatory protein, AcrR, involved in regulating virulence and motility. Here, we characterized a second LuxR-type regulator, AclR, in the group II strain Aac-5 of A. citrulli by mutagenesis, virulence and motility assays, and transcriptomic analysis. Deletion of aclR resulted in impaired twitching and swimming motility and flagellar formation and diminished virulence but increased biofilm formation. Transcriptomic analysis revealed that 1,379 genes were differentially expressed in the aclR mutant strain, including 29 genes involved in flagellar assembly and 3 involved in pili formation, suggesting a regulatory role for AclR in multiple important biological functions of A. citrulli. Together, our results not only indicate that AclR plays a global role in transcriptional regulation in A. citrulli influencing motility, biofilm formation, and virulence but also provide perspective regarding the regulatory network of biological functions in A. citrulli.[Formula: see text] Copyright © 2021 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.


Asunto(s)
Comamonadaceae , Proteínas Represoras/fisiología , Transactivadores/fisiología , Transcriptoma , Comamonadaceae/genética , Proteínas Represoras/genética , Transactivadores/genética , Transcriptoma/genética , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA